Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Genetic engineering of T cells for immunotherapy

Abstract

Genetically engineered T cell immunotherapies have provided remarkable clinical success to treat B cell acute lymphoblastic leukaemia by harnessing a patient’s own T cells to kill cancer, and these approaches have the potential to provide therapeutic benefit for numerous other cancers, infectious diseases and autoimmunity. By introduction of either a transgenic T cell receptor or a chimeric antigen receptor, T cells can be programmed to target cancer cells. However, initial studies have made it clear that the field will need to implement more complex levels of genetic regulation of engineered T cells to ensure both safety and efficacy. Here, we review the principles by which our knowledge of genetics and genome engineering will drive the next generation of adoptive T cell therapies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Autologous and allogeneic T cell immunotherapy.
Fig. 2: The genetic outcome of modifying the T cell genome.
Fig. 3: Gene-engineered T cell products to enhance efficacy.
Fig. 4: Addition of armour or subtraction of suppressive genes or their transcripts enables TME resistance.
Fig. 5: Toxicity risks associated with gene-engineered T cells.
Fig. 6: Gene-engineered T cell products for enhanced safety.

Similar content being viewed by others

References

  1. Rappuoli, R., Mandl, C. W., Black, S. & De Gregorio, E. Vaccines for the twenty-first century society. Nat. Rev. Immunol. 11, 865–872 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Sliwkowski, M. X. & Mellman, I. Antibody therapeutics in cancer. Science 341, 1192–1198 (2013).

    CAS  PubMed  Google Scholar 

  3. Pardoll, D. M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12, 252–264 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Feldmann, M. & Maini, R. N. Anti-TNF alpha therapy of rheumatoid arthritis: what have we learned? Annu. Rev. Immunol. 19, 163–196 (2001).

    CAS  PubMed  Google Scholar 

  5. Kumar, B. V., Connors, T. J. & Farber, D. L. Human T cell development, localization, and function throughout life. Immunity 48, 202–213 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Jameson, S. C. & Masopust, D. Understanding subset diversity in T cell memory. Immunity 48, 214–226 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Francica, J. R. et al. Steric shielding of surface epitopes and impaired immune recognition induced by the ebola virus glycoprotein. PLoS Pathog. 6, e1001098 (2010).

    PubMed  PubMed Central  Google Scholar 

  8. Wei, F. et al. Strength of PD-1 signaling differentially affects T-cell effector functions. Proc. Natl Acad. Sci. USA 110, E2480–E2489 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Park, J. H., Geyer, M. B. & Brentjens, R. J. CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date. Blood 127, 3312–3320 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Stadtmauer, E. A. et al. CRISPR-engineered T cells in patients with refractory cancer. Science 367, eaba7365 (2020). This is the first report on the in vivo fate and genomic ramifications of CRISPR-engineered TCR T cells in a phase I clinical trial.

    CAS  PubMed  Google Scholar 

  11. Li, J., Hong, S., Chen, W., Zuo, E. & Yang, H. Advances in detecting and reducing off-target effects generated by CRISPR-mediated genome editing. J. Genet. Genomics 46, 513–521 (2019).

    PubMed  Google Scholar 

  12. Maldini, C. R., Ellis, G. I. & Riley, J. L. CAR T cells for infection, autoimmunity and allotransplantation. Nat. Rev. Immunol. 18, 605–616 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Rosado-Sanchez, I. & Levings, M. K. Building a CAR-Treg: going from the basic to the luxury model. Cell Immunol. 358, 104220 (2020).

    CAS  PubMed  Google Scholar 

  14. Klichinsky, M. et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 38, 947–953 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Kimbrel, E. A. & Lanza, R. Next-generation stem cells - ushering in a new era of cell-based therapies. Nat. Rev. Drug Discov. 19, 463–479 (2020).

    CAS  PubMed  Google Scholar 

  16. Hartweger, H. et al. HIV-specific humoral immune responses by CRISPR/Cas9-edited B cells. J. Exp. Med. 216, 1301–1310 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Fesnak, A. D., June, C. H. & Levine, B. L. Engineered T cells: the promise and challenges of cancer immunotherapy. Nat. Rev. Cancer 16, 566–581 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Newrzela, S. et al. T-cell receptor diversity prevents T-cell lymphoma development. Leukemia 26, 2499–2507 (2012).

    CAS  PubMed  Google Scholar 

  19. Scholler, J. et al. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci. Transl Med. 4, 132ra153 (2012).

    Google Scholar 

  20. O’Leary, M. C. et al. FDA approval summary: tisagenlecleucel for treatment of patients with relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Clin. Cancer Res. 25, 1142–1146 (2019).

    PubMed  Google Scholar 

  21. Bouchkouj, N. et al. FDA approval summary: axicabtagene ciloleucel for relapsed or refractory large B-cell lymphoma. Clin. Cancer Res. 25, 1702–1708 (2019).

    PubMed  Google Scholar 

  22. Hirayama, A. V. et al. High rate of durable complete remission in follicular lymphoma after CD19 CAR-T cell immunotherapy. Blood 134, 636–640 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Schuster, S. J. et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N. Engl. J. Med. 380, 45–56 (2019).

    CAS  PubMed  Google Scholar 

  24. Maude, S. L. et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 378, 439–448 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Locke, F. L. et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial. Lancet Oncol. 20, 31–42 (2019).

    CAS  PubMed  Google Scholar 

  26. Fiorenza, S., Ritchie, D. S., Ramsey, S. D., Turtle, C. J. & Roth, J. A. Value and affordability of CAR T-cell therapy in the United States. Bone Marrow Transplant. 55, 1706–1715 (2020).

    PubMed  Google Scholar 

  27. Kagoya, Y. et al. Genetic ablation of HLA class I, class II, and the T-cell receptor enables allogeneic T cells to be used for adoptive T-cell therapy. Cancer Immunol. Res. 8, 926–936 (2020).

    CAS  PubMed  Google Scholar 

  28. Cruz, C. R. et al. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood 122, 2965–2973 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Kochenderfer, J. N. et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 122, 4129–4139 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Huls, M. H. et al. Clinical application of Sleeping Beauty and artificial antigen presenting cells to genetically modify T cells from peripheral and umbilical cord blood. J. Vis. Exp. https://doi.org/10.3791/50070 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Pegram, H. J. et al. IL-12-secreting CD19-targeted cord blood-derived T cells for the immunotherapy of B-cell acute lymphoblastic leukemia. Leukemia 29, 415–422 (2015).

    CAS  PubMed  Google Scholar 

  32. Ueda, T. & Kaneko, S. In vitro differentiation of T cell: from CAR-modified T-iPSC. Methods Mol. Biol. 2048, 85–91 (2019).

    CAS  PubMed  Google Scholar 

  33. Themeli, M. et al. Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy. Nat. Biotechnol. 31, 928–933 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Yan, L. Z. et al. Allogeneic CAR-T for treatment of relapsed and/or refractory multiple myeloma: four cases report and literatures review. Zhonghua Xue Ye Xue Za Zhi 40, 650–655 (2019).

    CAS  PubMed  Google Scholar 

  35. Brudno, J. N. et al. Allogeneic T cells that express an anti-CD19 chimeric antigen receptor induce remissions of B-cell malignancies that progress after allogeneic hematopoietic stem-cell transplantation without causing graft-versus-host disease. J. Clin. Oncol. 34, 1112–1121 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Eapen, M. et al. Effect of graft source on unrelated donor haemopoietic stem-cell transplantation in adults with acute leukaemia: a retrospective analysis. Lancet Oncol. 11, 653–660 (2010).

    PubMed  PubMed Central  Google Scholar 

  37. Kwoczek, J. et al. Cord blood-derived T cells allow the generation of a more naive tumor-reactive cytotoxic T-cell phenotype. Transfusion 58, 88–99 (2018).

    CAS  PubMed  Google Scholar 

  38. Torikai, H. et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood 119, 5697–5705 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Stenger, D. et al. Endogenous TCR promotes in vivo persistence of CD19-CAR-T cells compared to a CRISPR/Cas9-mediated TCR knockout CAR. Blood 136, 1407–1418 (2020).

    PubMed  Google Scholar 

  40. Steentoft, C. et al. Glycan-directed CAR-T cells. Glycobiology 28, 656–669 (2018).

    CAS  PubMed  Google Scholar 

  41. Hughes, M. S. et al. Transfer of a TCR gene derived from a patient with a marked antitumor response conveys highly active T-cell effector functions. Hum. Gene Ther. 16, 457–472 (2005).

    CAS  PubMed  Google Scholar 

  42. Zhao, Y. et al. Primary human lymphocytes transduced with NY-ESO-1 antigen-specific TCR genes recognize and kill diverse human tumor cell lines. J. Immunol. 174, 4415–4423 (2005).

    CAS  PubMed  Google Scholar 

  43. Ahmadi, M. et al. CD3 limits the efficacy of TCR gene therapy in vivo. Blood 118, 3528–3537 (2011).

    CAS  PubMed  Google Scholar 

  44. van Loenen, M. M. et al. Mixed T cell receptor dimers harbor potentially harmful neoreactivity. Proc. Natl Acad. Sci. USA 107, 10972–10977 (2010).

    PubMed  PubMed Central  Google Scholar 

  45. Shao, H. et al. TCR mispairing in genetically modified T cells was detected by fluorescence resonance energy transfer. Mol. Biol. Rep. 37, 3951–3956 (2010).

    CAS  PubMed  Google Scholar 

  46. Cohen, C. J. et al. Enhanced antitumor activity of T cells engineered to express T-cell receptors with a second disulfide bond. Cancer Res. 67, 3898–3903 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Bendle, G. M. et al. Lethal graft-versus-host disease in mouse models of T cell receptor gene therapy. Nat. Med. 16, 565–570 (2010).

    CAS  PubMed  Google Scholar 

  48. Govers, C., Sebestyen, Z., Coccoris, M., Willemsen, R. A. & Debets, R. T cell receptor gene therapy: strategies for optimizing transgenic TCR pairing. Trends Mol. Med. 16, 77–87 (2010).

    CAS  PubMed  Google Scholar 

  49. Yang, S. et al. Development of optimal bicistronic lentiviral vectors facilitates high-level TCR gene expression and robust tumor cell recognition. Gene Ther. 15, 1411–1423 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Legut, M., Dolton, G., Mian, A. A., Ottmann, O. G. & Sewell, A. K. CRISPR-mediated TCR replacement generates superior anticancer transgenic T cells. Blood 131, 311–322 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Okamoto, S. et al. Improved expression and reactivity of transduced tumor-specific TCRs in human lymphocytes by specific silencing of endogenous TCR. Cancer Res. 69, 9003–9011 (2009).

    CAS  PubMed  Google Scholar 

  52. Robbins, P. F. et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J. Clin. Oncol. 29, 917–924 (2011).

    PubMed  PubMed Central  Google Scholar 

  53. Maldini, C. R. et al. Dual CD4-based CAR T cells with distinct costimulatory domains mitigate HIV pathogenesis in vivo. Nat. Med. 26, 1776–1787 (2020).

    PubMed  Google Scholar 

  54. Guedan, S. et al. ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cells. Blood 124, 1070–1080 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Kawalekar, O. U. et al. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity 44, 380–390 (2016).

    CAS  PubMed  Google Scholar 

  56. Liu, X. et al. Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice. Cancer Res. 75, 3596–3607 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Ghorashian, S. et al. Enhanced CAR T cell expansion and prolonged persistence in pediatric patients with ALL treated with a low-affinity CD19 CAR. Nat. Med. 25, 1408–1414 (2019).

    CAS  PubMed  Google Scholar 

  58. Ellebrecht, C. T. et al. Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease. Science 353, 179–184 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Brusko, T. M., Wasserfall, C. H., Clare-Salzler, M. J., Schatz, D. A. & Atkinson, M. A. Functional defects and the influence of age on the frequency of CD4+ CD25+ T-cells in type 1 diabetes. Diabetes 54, 1407–1414 (2005).

    CAS  PubMed  Google Scholar 

  60. Thiruppathi, M. et al. Functional defect in regulatory T cells in myasthenia gravis. Ann. N. Y. Acad. Sci. 1274, 68–76 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Viglietta, V., Baecher-Allan, C., Weiner, H. L. & Hafler, D. A. Loss of functional suppression by CD4+CD25+ regulatory T cells in patients with multiple sclerosis. J. Exp. Med. 199, 971–979 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Brunstein, C. G. et al. Umbilical cord blood-derived T regulatory cells to prevent GVHD: kinetics, toxicity profile, and clinical effect. Blood 127, 1044–1051 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Plesa, G. et al. TCR affinity and specificity requirements for human regulatory T-cell function. Blood 119, 3420–3430 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Boardman, D. A. et al. Expression of a chimeric antigen receptor specific for donor HLA class I enhances the potency of human regulatory T cells in preventing human skin transplant rejection. Am. J. Transpl. 17, 931–943 (2017).

    CAS  Google Scholar 

  65. MacDonald, K. G. et al. Alloantigen-specific regulatory T cells generated with a chimeric antigen receptor. J. Clin. Invest. 126, 1413–1424 (2016). This study demonstrates that human CAR Treg cells can prevent GVHD in vivo.

    PubMed  PubMed Central  Google Scholar 

  66. Noyan, F. et al. Prevention of allograft rejection by use of regulatory T cells with an MHC-specific chimeric antigen receptor. Am. J. Transpl. 17, 917–930 (2017).

    CAS  Google Scholar 

  67. Dawson, N. A. et al. Systematic testing and specificity mapping of alloantigen-specific chimeric antigen receptors in regulatory T cells. JCI Insight 4, e123672 (2019).

    PubMed Central  Google Scholar 

  68. Yeh, W. I. et al. Avidity and bystander suppressive capacity of human regulatory T cells expressing de novo autoreactive T-cell receptors in type 1 diabetes. Front. Immunol. 8, 1313 (2017).

    PubMed  PubMed Central  Google Scholar 

  69. Hull, C. M. et al. Generation of human islet-specific regulatory T cells by TCR gene transfer. J. Autoimmun. 79, 63–73 (2017).

    CAS  PubMed  Google Scholar 

  70. Dawson, N. A. J. et al. Functional effects of chimeric antigen receptor co-receptor signaling domains in human regulatory T cells. Sci. Transl Med. 12, eaaz3866 (2020).

    CAS  PubMed  Google Scholar 

  71. Boroughs, A. C. et al. Chimeric antigen receptor costimulation domains modulate human regulatory T cell function. JCI Insight 5, e126194 (2019).

    Google Scholar 

  72. Hippen, K. L. et al. Massive ex vivo expansion of human natural regulatory T cells (Tregs) with minimal loss of in vivo functional activity. Sci. Transl Med. 3, 83ra41 (2011).

    PubMed  PubMed Central  Google Scholar 

  73. McKenna, D. H., Jr. et al. Optimization of cGMP purification and expansion of umbilical cord blood-derived T-regulatory cells in support of first-in-human clinical trials. Cytotherapy 19, 250–262 (2017).

    CAS  PubMed  Google Scholar 

  74. Zhou, X. et al. Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat. Immunol. 10, 1000–1007 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Honaker, Y. et al. Gene editing to induce FOXP3 expression in human CD4+ T cells leads to a stable regulatory phenotype and function. Sci. Transl Med. 12, eaay6422 (2020).

    CAS  PubMed  Google Scholar 

  76. Kumar, M., Keller, B., Makalou, N. & Sutton, R. E. Systematic determination of the packaging limit of lentiviral vectors. Hum. Gene Ther. 12, 1893–1905 (2001).

    CAS  PubMed  Google Scholar 

  77. Mitchell, R. S. et al. Retroviral DNA integration: ASLV, HIV, and MLV show distinct target site preferences. PLoS Biol. 2, E234 (2004).

    PubMed  PubMed Central  Google Scholar 

  78. Yang, O. O. et al. Lysis of HIV-1-infected cells and inhibition of viral replication by universal receptor T cells. Proc. Natl Acad. Sci. USA 94, 11478–11483 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Deeks, S. G. et al. A phase II randomized study of HIV-specific T-cell gene therapy in subjects with undetectable plasma viremia on combination antiretroviral therapy. Mol. Ther. 5, 788–797 (2002).

    CAS  PubMed  Google Scholar 

  80. Mitsuyasu, R. T. et al. Prolonged survival and tissue trafficking following adoptive transfer of CD4zeta gene-modified autologous CD4+ and CD8+ T cells in human immunodeficiency virus-infected subjects. Blood 96, 785–793 (2000).

    CAS  PubMed  Google Scholar 

  81. Leibman, R. S. et al. Supraphysiologic control over HIV-1 replication mediated by CD8 T cells expressing a re-engineered CD4-based chimeric antigen receptor. PLoS Pathog. 13, e1006613 (2017).

    PubMed  PubMed Central  Google Scholar 

  82. Bobisse, S. et al. Reprogramming T lymphocytes for melanoma adoptive immunotherapy by T-cell receptor gene transfer with lentiviral vectors. Cancer Res. 69, 9385–9394 (2009).

    CAS  PubMed  Google Scholar 

  83. Zufferey, R., Donello, J. E., Trono, D. & Hope, T. J. Woodchuck hepatitis virus posttranscriptional regulatory element enhances expression of transgenes delivered by retroviral vectors. J. Virol. 73, 2886–2892 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Norrman, K. et al. Quantitative comparison of constitutive promoters in human ES cells. PLoS ONE 5, e12413 (2010).

    PubMed  PubMed Central  Google Scholar 

  85. Eyquem, J. et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 543, 113–117 (2017). The authors improve CAR function by CRISPR-mediated insertion of CAR into the TRAC gene, improving expression kinetics and decreasing exhaustion.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Masiuk, K. E., Laborada, J., Roncarolo, M. G., Hollis, R. P. & Kohn, D. B. Lentiviral gene therapy in HSCs restores lineage-specific Foxp3 expression and suppresses autoimmunity in a mouse model of IPEX syndrome. Cell Stem Cell 24, 309–317.e7 (2019).

    CAS  PubMed  Google Scholar 

  87. Fraietta, J. A. et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature 558, 307–312 (2018). Random insertion of CAR into the TET2 locus highlights the potential for targeting the epigenome to improve CAR T cell function and the value of analysis of the clinical product.

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Zhao, Y. et al. High-efficiency transfection of primary human and mouse T lymphocytes using RNA electroporation. Mol. Ther. 13, 151–159 (2006).

    CAS  PubMed  Google Scholar 

  89. Zhao, Y. et al. Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. Cancer Res. 70, 9053–9061 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Jin, Z. et al. The hyperactive sleeping beauty transposase SB100X improves the genetic modification of T cells to express a chimeric antigen receptor. Gene Ther. 18, 849–856 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Chicaybam, L. et al. Transposon-mediated generation of CAR-T cells shows efficient anti B-cell leukemia response after ex vivo expansion. Gene Ther. 27, 85–95 (2020).

    CAS  PubMed  Google Scholar 

  92. Gogol-Doring, A. et al. Genome-wide profiling reveals remarkable parallels between insertion site selection properties of the MLV retrovirus and the Piggybac transposon in primary human CD4+ T cells. Mol. Ther. 24, 592–606 (2016).

    PubMed  PubMed Central  Google Scholar 

  93. Berry, C., Hannenhalli, S., Leipzig, J. & Bushman, F. D. Selection of target sites for mobile DNA integration in the human genome. PLoS Comput. Biol. 2, e157 (2006).

    PubMed  PubMed Central  Google Scholar 

  94. Chicaybam, L. et al. CAR T cells generated using Sleeping Beauty transposon vectors and expanded with an EBV-transformed lymphoblastoid cell line display antitumor activity in vitro and in vivo. Hum. Gene Ther. 30, 511–522 (2019).

    CAS  PubMed  Google Scholar 

  95. Voigt, F. et al. Sleeping Beauty transposase structure allows rational design of hyperactive variants for genetic engineering. Nat. Commun. 7, 11126 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Querques, I. et al. A highly soluble Sleeping Beauty transposase improves control of gene insertion. Nat. Biotechnol. 37, 1502–1512 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Monjezi, R. et al. Enhanced CAR T-cell engineering using non-viral Sleeping Beauty transposition from minicircle vectors. Leukemia 31, 186–194 (2017).

    CAS  PubMed  Google Scholar 

  98. Kebriaei, P. et al. Phase I trials using sleeping beauty to generate CD19-specific CAR T cells. J. Clin. Invest. 126, 3363–3376 (2016).

    PubMed  PubMed Central  Google Scholar 

  99. Magnani, C. F. et al. Sleeping Beauty-engineered CAR T cells achieve antileukemic activity without severe toxicities. J. Clin. Invest. 130, 6021–6033 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Wang, J. et al. Highly efficient homology-driven genome editing in human T cells by combining zinc-finger nuclease mRNA and AAV6 donor delivery. Nucleic Acids Res. 44, e30 (2016).

    PubMed  Google Scholar 

  101. MacLeod, D. T. et al. Integration of a CD19 CAR into the TCR alpha chain locus streamlines production of allogeneic gene-edited CAR T cells. Mol. Ther. 25, 949–961 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Hubbard, N. et al. Targeted gene editing restores regulated CD40L function in X-linked hyper-IgM syndrome. Blood 127, 2513–2522 (2016).

    CAS  PubMed  Google Scholar 

  103. Sather, B. D. et al. Efficient modification of CCR5 in primary human hematopoietic cells using a megaTAL nuclease and AAV donor template. Sci. Transl Med. 7, 307ra156 (2015).

    PubMed  PubMed Central  Google Scholar 

  104. Roth, T. L. et al. Reprogramming human T cell function and specificity with non-viral genome targeting. Nature 559, 405–409 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Nguyen, D. N. et al. Polymer-stabilized Cas9 nanoparticles and modified repair templates increase genome editing efficiency. Nat. Biotechnol. 38, 44–49 (2020).

    CAS  PubMed  Google Scholar 

  106. Sadelain, M., Papapetrou, E. P. & Bushman, F. D. Safe harbours for the integration of new DNA in the human genome. Nat. Rev. Cancer 12, 51–58 (2011).

    PubMed  Google Scholar 

  107. Hale, M. et al. Engineering HIV-resistant, anti-HIV chimeric antigen receptor T cells. Mol. Ther. 25, 570–579 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Park, J. H. et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 378, 449–459 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Milone, M. C. et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol. Ther. 17, 1453–1464 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Morgan, P. et al. Can the flow of medicines be improved? Fundamental pharmacokinetic and pharmacological principles toward improving phase II survival. Drug Discov. Today 17, 419–424 (2012).

    CAS  PubMed  Google Scholar 

  111. Ager, A., Watson, H. A., Wehenkel, S. C. & Mohammed, R. N. Homing to solid cancers: a vascular checkpoint in adoptive cell therapy using CAR T-cells. Biochem. Soc. Trans. 44, 377–385 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Buckanovich, R. J. et al. Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Nat. Med. 14, 28–36 (2008).

    CAS  PubMed  Google Scholar 

  113. Motz, G. T. et al. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat. Med. 20, 607–615 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Yamamoto, T. N. et al. T cells genetically engineered to overcome death signaling enhance adoptive cancer immunotherapy. J. Clin. Invest. 129, 1551–1565 (2019).

    PubMed  PubMed Central  Google Scholar 

  115. Lu, P., Weaver, V. M. & Werb, Z. The extracellular matrix: a dynamic niche in cancer progression. J. Cell Biol. 196, 395–406 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Griffith, K. D. et al. In vivo distribution of adoptively transferred indium-111-labeled tumor infiltrating lymphocytes and peripheral blood lymphocytes in patients with metastatic melanoma. J. Natl Cancer Inst. 81, 1709–1717 (1989).

    CAS  PubMed  Google Scholar 

  117. Moon, E. K. et al. Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumors. Clin. Cancer Res. 20, 4262–4273 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Moon, E. K. et al. Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor. Clin. Cancer Res. 17, 4719–4730 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Caruana, I. et al. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat. Med. 21, 524–529 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Swanton, C. Intratumor heterogeneity: evolution through space and time. Cancer Res. 72, 4875–4882 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Sotillo, E. et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 5, 1282–1295 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. O’Rourke, D. M. et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl Med. 9, eaaa0984 (2017).

    PubMed  PubMed Central  Google Scholar 

  123. Rapoport, A. P. et al. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat. Med. 21, 914–921 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Feng, K. C. et al. Cocktail treatment with EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells in a patient with advanced cholangiocarcinoma. J. Hematol. Oncol. 10, 4 (2017).

    PubMed  PubMed Central  Google Scholar 

  125. Ruella, M. et al. Dual CD19 and CD123 targeting prevents antigen-loss relapses after CD19-directed immunotherapies. J. Clin. Invest. 126, 3814–3826 (2016).

    PubMed  PubMed Central  Google Scholar 

  126. Simon, B., Harrer, D. C., Schuler-Thurner, B., Schuler, G. & Uslu, U. Arming T cells with a gp100-specific TCR and a CSPG4-specific CAR using combined DNA- and RNA-based receptor transfer. Cancers 11, 696 (2019).

    CAS  PubMed Central  Google Scholar 

  127. Hegde, M. et al. Tandem CAR T cells targeting HER2 and IL13Ralpha2 mitigate tumor antigen escape. J. Clin. Invest. 126, 3036–3052 (2016).

    PubMed  PubMed Central  Google Scholar 

  128. Zah, E. et al. Systematically optimized BCMA/CS1 bispecific CAR-T cells robustly control heterogeneous multiple myeloma. Nat. Commun. 11, 2283 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. de Larrea, C. F. et al. Defining an optimal dual-targeted CAR T-cell therapy approach simultaneously targeting BCMA and GPRC5D to prevent BCMA escape-driven relapse in multiple myeloma. Blood Cancer Discov. 1, 146–154 (2020).

    Google Scholar 

  130. Han, X., Wang, Y., Wei, J. & Han, W. Multi-antigen-targeted chimeric antigen receptor T cells for cancer therapy. J. Hematol. Oncol. 12, 128 (2019).

    PubMed  PubMed Central  Google Scholar 

  131. Bielamowicz, K. et al. Trivalent CAR T cells overcome interpatient antigenic variability in glioblastoma. Neuro Oncol. 20, 506–518 (2018).

    CAS  PubMed  Google Scholar 

  132. Kudo, K. et al. T lymphocytes expressing a CD16 signaling receptor exert antibody-dependent cancer cell killing. Cancer Res. 74, 93–103 (2014).

    CAS  PubMed  Google Scholar 

  133. Landgraf, K. E. et al. ConvertibleCARs: a chimeric antigen receptor system for flexible control of activity and antigen targeting. Commun. Biol. 3, 296 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Minutolo, N. G., Hollander, E. E. & Powell, D. J., Jr. The emergence of universal immune receptor T cell therapy for cancer. Front. Oncol. 9, 176 (2019).

    PubMed  PubMed Central  Google Scholar 

  135. Minutolo, N. G. et al. Quantitative control of gene-engineered T-cell activity through the covalent attachment of targeting ligands to a universal immune receptor. J. Am. Chem. Soc. 142, 6554–6568 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Adachi, K. et al. IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat. Biotechnol. 36, 346–351 (2018).

    CAS  PubMed  Google Scholar 

  137. Luo, H. et al. Coexpression of IL7 and CCL21 increases efficacy of CAR-T cells in solid tumors without requiring preconditioned lymphodepletion. Clin. Cancer Res. 26, 5494–5505 (2020).

    CAS  PubMed  Google Scholar 

  138. Wang, N. et al. Efficacy and safety of CAR19/22 T-cell cocktail therapy in patients with refractory/relapsed B-cell malignancies. Blood 135, 17–27 (2020).

    PubMed  Google Scholar 

  139. Yan, Z. et al. A combination of humanised anti-CD19 and anti-BCMA CAR T cells in patients with relapsed or refractory multiple myeloma: a single-arm, phase 2 trial. Lancet Haematol. 6, e521–e529 (2019).

    PubMed  Google Scholar 

  140. Zhao, W. H. et al. A phase 1, open-label study of LCAR-B38M, a chimeric antigen receptor T cell therapy directed against B cell maturation antigen, in patients with relapsed or refractory multiple myeloma. J. Hematol. Oncol. 11, 141 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Lawrence, M. S. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Ecker, C. & Riley, J. L. Translating in vitro T cell metabolic findings to in vivo tumor models of nutrient competition. Cell Metab. 28, 190–195 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Munn, D. H. & Bronte, V. Immune suppressive mechanisms in the tumor microenvironment. Curr. Opin. Immunol. 39, 1–6 (2016).

    CAS  PubMed  Google Scholar 

  144. Lu, Y. et al. Safety and feasibility of CRISPR-edited T cells in patients with refractory non-small-cell lung cancer. Nat. Med. 26, 732–740 (2020).

    CAS  PubMed  Google Scholar 

  145. Neuzillet, C. et al. Targeting the TGFbeta pathway for cancer therapy. Pharmacol. Ther. 147, 22–31 (2015).

    CAS  PubMed  Google Scholar 

  146. Kloss, C. C. et al. Dominant-negative TGF-beta receptor enhances PSMA-targeted human CAR T cell proliferation and augments prostate cancer eradication. Mol. Ther. 26, 1855–1866 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Powell, A. B. et al. Medulloblastoma rendered susceptible to NK-cell attack by TGFbeta neutralization. J. Transl. Med. 17, 321 (2019).

    PubMed  PubMed Central  Google Scholar 

  148. Bollard, C. M. et al. Tumor-specific T-cells engineered to overcome tumor immune evasion induce clinical responses in patients with relapsed hodgkin lymphoma. J. Clin. Oncol. 36, 1128–1139 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Beavis, P. A. et al. Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy. J. Clin. Invest. 127, 929–941 (2017).

    PubMed  PubMed Central  Google Scholar 

  150. Sim, G. C. et al. IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients. J. Clin. Invest. 124, 99–110 (2014).

    CAS  PubMed  Google Scholar 

  151. Suryadevara, C. M. et al. Preventing Lck activation in CAR T cells confers Treg resistance but requires 4-1BB signaling for them to persist and treat solid tumors in nonlymphodepleted hosts. Clin. Cancer Res. 25, 358–368 (2019).

    CAS  PubMed  Google Scholar 

  152. Franchina, D. G., He, F. & Brenner, D. Survival of the fittest: cancer challenges T cell metabolism. Cancer Lett. 412, 216–223 (2018).

    CAS  PubMed  Google Scholar 

  153. Kawalekar, O. U. et al. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T Cells. Immunity 44, 712 (2016).

    CAS  PubMed  Google Scholar 

  154. Fultang, L. et al. Metabolic engineering against the arginine microenvironment enhances CAR-T cell proliferation and therapeutic activity. Blood 136, 1155–1160 (2020).

    PubMed  PubMed Central  Google Scholar 

  155. Leonard, J. P. et al. Effects of single-dose interleukin-12 exposure on interleukin-12-associated toxicity and interferon-gamma production. Blood 90, 2541–2548 (1997).

    CAS  PubMed  Google Scholar 

  156. Zhang, L. et al. Improving adoptive T cell therapy by targeting and controlling IL-12 expression to the tumor environment. Mol. Ther. 19, 751–759 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Joung, J. et al. Genome-scale CRISPR-Cas9 knockout and transcriptional activation screening. Nat. Protoc. 12, 828–863 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Shifrut, E. et al. Genome-wide CRISPR screens in primary human T cells reveal key regulators of immune function. Cell 175, 1958–1971.e15 (2018). This study uses a genome-wide CRISPR screen to unearth proteins that limit T cell proliferation and killing in the tumour microenvironment.

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Cortez, J. T. et al. CRISPR screen in regulatory T cells reveals modulators of Foxp3. Nature 582, 416–420 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Dong, M. B. et al. Systematic immunotherapy target discovery using genome-scale in vivo CRISPR screens in CD8 T cells. Cell 178, 1189–1204.e23 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Roth, T. L. et al. Pooled knockin targeting for genome engineering of cellular immunotherapies. Cell 181, 728–744.e21 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Wei, J. et al. PD-1 silencing impairs the anti-tumor function of chimeric antigen receptor modified T cells by inhibiting proliferation activity. J. Immunother. Cancer 7, 209 (2019).

    PubMed  PubMed Central  Google Scholar 

  163. Cherkassky, L. et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J. Clin. Invest. 126, 3130–3144 (2016).

    PubMed  PubMed Central  Google Scholar 

  164. Ankri, C., Shamalov, K., Horovitz-Fried, M., Mauer, S. & Cohen, C. J. Human T cells engineered to express a programmed death 1/28 costimulatory retargeting molecule display enhanced antitumor activity. J. Immunol. 191, 4121–4129 (2013).

    CAS  PubMed  Google Scholar 

  165. Parry, R. V. et al. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell Biol. 25, 9543–9553 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Rafiq, S. et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat. Biotechnol. 36, 847–856 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Zhang, A. et al. Secretion of human soluble programmed cell death protein 1 by chimeric antigen receptor-modified T cells enhances anti-tumor efficacy. Cytotherapy 22, 734–743 (2020).

    CAS  PubMed  Google Scholar 

  168. Chang, Z. L., Silver, P. A. & Chen, Y. Y. Identification and selective expansion of functionally superior T cells expressing chimeric antigen receptors. J. Transl. Med. 13, 161 (2015).

    PubMed  PubMed Central  Google Scholar 

  169. Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Chen, Z. et al. TCF-1-centered transcriptional network drives an effector versus exhausted CD8 T cell-fate decision. Immunity 51, 840–855.e5 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Chen, J. et al. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 567, 530–534 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Odorizzi, P. M., Pauken, K. E., Paley, M. A., Sharpe, A. & Wherry, E. J. Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells. J. Exp. Med. 212, 1125–1137 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Greenbaum, U. et al. Chimeric antigen receptor T-cell therapy toxicities. Br. J. Clin. Pharmacol. https://doi.org/10.1111/bcp.14403 (2020).

    Article  PubMed  Google Scholar 

  174. Curran, K. J. et al. Toxicity and response after CD19-specific CAR T-cell therapy in pediatric/young adult relapsed/refractory B-ALL. Blood 134, 2361–2368 (2019).

    PubMed  PubMed Central  Google Scholar 

  175. Neelapu, S. S. et al. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat. Rev. Clin. Oncol. 15, 47–62 (2018).

    CAS  PubMed  Google Scholar 

  176. Howard, S. C., Trifilio, S., Gregory, T. K., Baxter, N. & McBride, A. Tumor lysis syndrome in the era of novel and targeted agents in patients with hematologic malignancies: a systematic review. Ann. Hematol. 95, 563–573 (2016).

    CAS  PubMed  Google Scholar 

  177. Fried, S. et al. Early and late hematologic toxicity following CD19 CAR-T cells. Bone Marrow Transpl. 54, 1643–1650 (2019).

    CAS  Google Scholar 

  178. Teachey, D. T. et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov. 6, 664–679 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Ruella, M. et al. Induction of resistance to chimeric antigen receptor T cell therapy by transduction of a single leukemic B cell. Nat. Med. 24, 1499–1503 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Hacein-Bey-Abina, S. et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 302, 415–419 (2003).

    CAS  PubMed  Google Scholar 

  181. Marcucci, K. T. et al. Retroviral and lentiviral safety analysis of gene-modified T cell products and infused HIV and oncology patients. Mol. Ther. 26, 269–279 (2018).

    CAS  PubMed  Google Scholar 

  182. Morgan, R. A. et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J. Immunother. 36, 133–151 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Beatty, G. L. et al. Activity of mesothelin-specific chimeric antigen receptor T cells against pancreatic carcinoma metastases in a phase 1 trial. Gastroenterology 155, 29–32 (2018).

    CAS  PubMed  Google Scholar 

  184. Maus, M. V. et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol. Res. 1, 26–31 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Tasian, S. K. et al. Optimized depletion of chimeric antigen receptor T cells in murine xenograft models of human acute myeloid leukemia. Blood 129, 2395–2407 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Jonnalagadda, M. et al. Efficient selection of genetically modified human T cells using methotrexate-resistant human dihydrofolate reductase. Gene Ther. 20, 853–860 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Cavaco, M., Gaspar, D., Arb Castanho, M. & Neves, V. Antibodies for the treatment of brain metastases, a dream or a reality? Pharmaceutics 12, 62 (2020).

    CAS  PubMed Central  Google Scholar 

  188. Gargett, T. & Brown, M. P. The inducible caspase-9 suicide gene system as a “safety switch” to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Front. Pharmacol. 5, 235 (2014).

    PubMed  PubMed Central  Google Scholar 

  189. Gu, X., He, D., Li, C., Wang, H. & Yang, G. Development of inducible CD19-CAR T cells with a Tet-on system for controlled activity and enhanced clinical safety. Int. J. Mol. Sci. 19, 3455 (2018).

    PubMed Central  Google Scholar 

  190. Drent, E. et al. Feasibility of controlling CD38-CAR T cell activity with a Tet-on inducible CAR design. PLoS ONE 13, e0197349 (2018).

    PubMed  PubMed Central  Google Scholar 

  191. Sakemura, R. et al. A Tet-on inducible system for controlling CD19-chimeric antigen receptor expression upon drug administration. Cancer Immunol. Res. 4, 658–668 (2016).

    CAS  PubMed  Google Scholar 

  192. Zhang, R. Y. et al. Doxycycline inducible chimeric antigen receptor T cells targeting CD147 for hepatocellular carcinoma therapy. Front. Cell Dev. Biol. 7, 233 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Foster, A. E. et al. Regulated expansion and survival of chimeric antigen receptor-modified T cells using small molecule-dependent inducible MyD88/CD40. Mol. Ther. 25, 2176–2188 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Duong, M. T. et al. Two-dimensional regulation of CAR-T cell therapy with orthogonal switches. Mol. Ther. Oncolytics 12, 124–137 (2019).

    PubMed  Google Scholar 

  195. Wu, C. Y., Roybal, K. T., Puchner, E. M., Onuffer, J. & Lim, W. A. Remote control of therapeutic T cells through a small molecule-gated chimeric receptor. Science 350, aab4077 (2015).

    PubMed  PubMed Central  Google Scholar 

  196. Huang, Z. et al. Engineering light-controllable CAR T cells for cancer immunotherapy. Sci. Adv. 6, eaay9209 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Lee, S. M. et al. A Chemical switch system to modulate chimeric antigen receptor T cell activity through proteolysis-targeting chimaera technology. ACS Synth. Biol. 9, 987–992 (2020).

    CAS  PubMed  Google Scholar 

  198. Juillerat, A. et al. Modulation of chimeric antigen receptor surface expression by a small molecule switch. BMC Biotechnol. 19, 44 (2019).

    PubMed  PubMed Central  Google Scholar 

  199. Richman, S. A. et al. Ligand-induced degradation of a CAR permits reversible remote control of CAR T cell activity in vitro and in vivo. Mol. Ther. 28, 1600–1613 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Mestermann, K. et al. The tyrosine kinase inhibitor dasatinib acts as a pharmacologic on/off switch for CAR T cells. Sci. Transl Med. 11, eaau5907 (2019). This article reports a pharmacological approach to modulating T cell function in vivo.

    PubMed  PubMed Central  Google Scholar 

  201. Ebert, L. M., Yu, W., Gargett, T. & Brown, M. P. Logic-gated approaches to extend the utility of chimeric antigen receptor T-cell technology. Biochem. Soc. Trans. 46, 391–401 (2018).

    CAS  PubMed  Google Scholar 

  202. Zah, E., Lin, M. Y., Silva-Benedict, A., Jensen, M. C. & Chen, Y. Y. T cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B Cells. Cancer Immunol. Res. 4, 498–508 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Fedorov, V. D., Themeli, M. & Sadelain, M. PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci. Transl Med. 5, 215ra172 (2013).

    PubMed  PubMed Central  Google Scholar 

  204. Lajoie, M. J. et al. Designed protein logic to target cells with precise combinations of surface antigens. Science 369, 1637–1643 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Srivastava, S. et al. Logic-gated ROR1 chimeric antigen receptor expression rescues T cell-mediated toxicity to normal tissues and enables selective tumor targeting. Cancer Cell 35, 489–503 e488 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Roybal, K. T. et al. Precision tumor recognition by T cells with combinatorial antigen-sensing circuits. Cell 164, 770–779 (2016). This study describes the development of synNotch CAR T cells operating on IF/THEN logic.

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Juillerat, A. et al. An oxygen sensitive self-decision making engineered CAR T-cell. Sci. Rep. 7, 39833 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  208. Michaels, Y. S. et al. Precise tuning of gene expression levels in mammalian cells. Nat. Commun. 10, 818 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  209. Chassin, H. et al. A modular degron library for synthetic circuits in mammalian cells. Nat. Commun. 10, 2013 (2019).

    PubMed  PubMed Central  Google Scholar 

  210. Ying, Z. et al. A safe and potent anti-CD19 CAR T cell therapy. Nat. Med. 25, 947–953 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  211. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019). This study describes the development of prime editing for genome editing in the absence of DNA double-strand breaks.

    CAS  PubMed  PubMed Central  Google Scholar 

  212. Tebas, P. et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N. Engl. J. Med. 370, 901–910 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Paschon, D. E. et al. Diversifying the structure of zinc finger nucleases for high-precision genome editing. Nat. Commun. 10, 1133 (2019).

    PubMed  PubMed Central  Google Scholar 

  214. Perez, E. E. et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat. Biotechnol. 26, 808–816 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Wilen, C. B. et al. Engineering HIV-resistant human CD4+ T cells with CXCR4-specific zinc-finger nucleases. PLoS Pathog. 7, e1002020 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  216. Yuan, J. et al. Zinc-finger nuclease editing of human cxcr4 promotes HIV-1 CD4+ T cell resistance and enrichment. Mol. Ther. 20, 849–859 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  217. Provasi, E. et al. Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer. Nat. Med. 18, 807–815 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Qasim, W. et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci. Transl Med. 9, eaaj2013 (2017).

    PubMed  Google Scholar 

  219. Valton, J. et al. A multidrug-resistant engineered CAR T cell for allogeneic combination immunotherapy. Mol. Ther. 23, 1507–1518 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  220. Mussolino, C. et al. A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic Acids Res. 39, 9283–9293 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  221. Sachdeva, M. et al. Repurposing endogenous immune pathways to tailor and control chimeric antigen receptor T cell functionality. Nat. Commun. 10, 5100 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  222. Ren, J. et al. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin. Cancer Res. 23, 2255–2266 (2017). The authors describe a CRISPR–Cas9-based triple knockout of genes encoding TCR, β2M and PD1 in primary human T cells.

    CAS  PubMed  Google Scholar 

  223. Liu, Z. et al. Genome editing of the HIV co-receptors CCR5 and CXCR4 by CRISPR-Cas9 protects CD4+ T cells from HIV-1 infection. Cell Biosci. 7, 47 (2017).

    PubMed  PubMed Central  Google Scholar 

  224. Anderson, W., Thorpe, J., Long, S. A. & Rawlings, D. J. Efficient CRISPR/Cas9 disruption of autoimmune-associated genes reveals key signaling programs in primary human T cells. J. Immunol. 203, 3166–3178 (2019).

    CAS  PubMed  Google Scholar 

  225. Gaudelli, N. M. et al. Directed evolution of adenine base editors with increased activity and therapeutic application. Nat. Biotechnol. 38, 892–900 (2020).

    CAS  PubMed  Google Scholar 

  226. Gornalusse, G. G. et al. HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells. Nat. Biotechnol. 35, 765–772 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  227. Mo, F. et al. Engineered off-the-shelf therapeutic T cells resist host immune rejection. Nat. Biotechnol. (2020).

  228. Wu, W. et al. Multiple signaling roles of CD3epsilon and its application in CAR-T cell therapy. Cell 182, 855–871.e23 (2020).

    CAS  PubMed  Google Scholar 

  229. Roberts, M. R. et al. Targeting of human immunodeficiency virus-infected cells by CD8+ T lymphocytes armed with universal T-cell receptors. Blood 84, 2878–2889 (1994).

    CAS  PubMed  Google Scholar 

  230. Maher, J., Brentjens, R. J., Gunset, G., Riviere, I. & Sadelain, M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor. Nat. Biotechnol. 20, 70–75 (2002).

    CAS  PubMed  Google Scholar 

  231. Porter, D. L., Levine, B. L., Kalos, M., Bagg, A. & June, C. H. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365, 725–733 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  232. Kochenderfer, J. N. et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 119, 2709–2720 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  233. Brentjens, R. J. et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci. Transl Med. 5, 177ra138 (2013).

    Google Scholar 

  234. Porter, D. L. et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci. Transl Med. 7, 303ra139 (2015).

    PubMed  PubMed Central  Google Scholar 

  235. Kim, G. B., Hege, K. & Riley, J. L. CAR talk: how cancer-specific CAR T cells can instruct how to build CAR T cells to cure HIV. Front. Immunol. 10, 2310 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  236. Maldini, C. R. et al. HIV-resistant and HIV-specific CAR-modified CD4+ T cells mitigate HIV disease progression and confer CD4+ T cell help in vivo. Mol. Ther. 28, 1585–1599 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  237. Herzig, E. et al. Attacking latent HIV with convertible CAR-T cells, a highly adaptable killing platform. Cell 179, 880–894.e10 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  238. Anthony-Gonda, K. et al. Multispecific anti-HIV duoCAR-T cells display broad in vitro antiviral activity and potent in vivo elimination of HIV-infected cells in a humanized mouse model. Sci. Transl Med. 11, eaav5685 (2019).

    PubMed  PubMed Central  Google Scholar 

  239. Ali, A. et al. HIV-1-specific chimeric antigen receptors based on broadly neutralizing antibodies. J. Virol. 90, 6999–7006 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  240. Festag, M. M. et al. Evaluation of a fully human, hepatitis b virus-specific chimeric antigen receptor in an immunocompetent mouse model. Mol. Ther. 27, 947–959 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  241. Sautto, G. A. et al. Chimeric antigen receptor (CAR)-engineered T cells redirected against hepatitis C virus (HCV) E2 glycoprotein. Gut 65, 512–523 (2016).

    CAS  PubMed  Google Scholar 

  242. Kumaresan, P. R. et al. Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection. Proc. Natl Acad. Sci. USA 111, 10660–10665 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  243. Bitinaite, J., Wah, D. A., Aggarwal, A. K. & Schildkraut, I. FokI dimerization is required for DNA cleavage. Proc. Natl Acad. Sci. USA 95, 10570–10575 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  244. Kim, Y. et al. A library of TAL effector nucleases spanning the human genome. Nat. Biotechnol. 31, 251–258 (2013).

    CAS  PubMed  Google Scholar 

  245. Boissel, S. et al. megaTALs: a rare-cleaving nuclease architecture for therapeutic genome engineering. Nucleic Acids Res. 42, 2591–2601 (2014).

    CAS  PubMed  Google Scholar 

  246. Osborn, M. J. et al. Evaluation of TCR gene editing achieved by TALENs, CRISPR/Cas9, and megaTAL nucleases. Mol. Ther. 24, 570–581 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  247. Qi, L. S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  248. Konermann, S. et al. Optical control of mammalian endogenous transcription and epigenetic states. Nature 500, 472–476 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  249. Yeo, N. C. et al. An enhanced CRISPR repressor for targeted mammalian gene regulation. Nat. Methods 15, 611–616 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  250. Huang, Y. H. et al. DNA epigenome editing using CRISPR-Cas SunTag-directed DNMT3A. Genome Biol. 18, 176 (2017).

    PubMed  PubMed Central  Google Scholar 

  251. Koonin, E. V., Makarova, K. S. & Zhang, F. Diversity, classification and evolution of CRISPR-Cas systems. Curr. Opin. Microbiol. 37, 67–78 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  252. Komor, A. C., Badran, A. H. & Liu, D. R. CRISPR-based technologies for the manipulation of eukaryotic genomes. Cell 168, 20–36 (2017).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors are grateful to the members of the Center for Cellular Immunotherapies for in-depth discussions regarding the topics discussed in this Review and apologize to the many investigators whose work they were unable to cite. The authors gratefully acknowledge funding from the NIH (U19AI117950, U19AI149680 UM1AI126620 and UG3DK122644), Helmsley Charitable Trust and Tmunity Therapeutics.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to James L. Riley.

Ethics declarations

Competing interests

J.L.R. is a co-founder and shareholder of Tmunity Therapeutics. He has been in receipt of research funding from Tmunity Therapeutics. N.C.S. is a shareholder of Tmunity Therapeutics and Fate Therapeutics. G.I.E. declares no competing interests.

Additional information

Peer review information

Nature Reviews Genetics thanks M. K. Levings, Z. Li, E. L. Smith, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

NCT01236573: https://clinicaltrials.gov/ct2/show/NCT01236573

NCT03240328: https://clinicaltrials.gov/ct2/show/NCT03240328

NCT03617198: https://clinicaltrials.gov/ct2/show/NCT03617198

NCT04422912: https://clinicaltrials.gov/ct2/show/NCT04422912

Glossary

Allogeneic

A term to denote a genetically different individual of the same species.

Antigen loss

A mechanism of cancer or pathogen recurrence in which the target (tumour or pathogen) escapes engineered T cell recognition, typically via exon skipping, downregulation or alteration of the target antigen.

Epitope spreading

Diversification of the immune response by endogenous immune cells against new targets following engineered T cell therapy.

Boolean logic gate

AND, OR and NOT functions, which can describe the response of combinations of immune receptors to multiple antigens.

Immune synapse

The location of physical interaction between an immune cell and its activator, either another immune cell or a non-haematopoietic target cell. The strength and the type of signals exchanged at the immune synapse can modulate an immune response.

Single-chain variable fragment

(scFv). A linear transposition of an antibody’s heavy and light chains separated by a flexible linker, which retains the antigen binding capacity of the original antibody and can be used as the binder domain of a chimeric antigen receptor molecule.

Monoclonal antibodies

(mAbs). Antibodies purified from a single B cell hybridoma that can be used as a diagnostic or therapeutic product.

Fc-dependent mechanisms

A series of effector modalities by which targets bound by antibodies are depleted via recognition of the constant region of the antibody molecule.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ellis, G.I., Sheppard, N.C. & Riley, J.L. Genetic engineering of T cells for immunotherapy. Nat Rev Genet 22, 427–447 (2021). https://doi.org/10.1038/s41576-021-00329-9

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-021-00329-9

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer